Anti-cancer agents in medicinal chemistry最新文献

筛选
英文 中文
Potentials of N-Acyl hydrazones Against Colorectal Cancer: A Mini Review. n -酰基腙类化合物抗结直肠癌的潜力综述
IF 2.6 4区 医学
Anti-cancer agents in medicinal chemistry Pub Date : 2025-01-23 DOI: 10.2174/0118715206356253241223040825
Darna Mounika, Sai Satya Sri Pulla, Swapnil Anil Sule, Vidya Jyothi Alli, Surender Singh Jadav
{"title":"Potentials of N-Acyl hydrazones Against Colorectal Cancer: A Mini Review.","authors":"Darna Mounika, Sai Satya Sri Pulla, Swapnil Anil Sule, Vidya Jyothi Alli, Surender Singh Jadav","doi":"10.2174/0118715206356253241223040825","DOIUrl":"https://doi.org/10.2174/0118715206356253241223040825","url":null,"abstract":"<p><p>Colorectal cancer (CRC) is a malignant gastrointestinal tract disorder with high occurrence and mortality index and showing an upsurge. Standard therapies for treating CRC are surgery and chemotherapy. Despite great effort in developing effective treatments, the progress is limited due to its relapse and recurrence. Prognosis of metastatic CRC is always complicated. This condition can be evaded by a novel approach i.e., targeted therapy which increases the survival rate in CRC patients by blocking important pathways and acting on immune checkpoints. Drugs with N-acyl hydrazones (NAH) are currently being employed treatment of infectious diseases and disorders. NAH in combination with diverse heterocycles, natural product isolates are identified as interesting CRC inhibitors under-explored. This review provides an overview of the existing CRC targeted compounds having acyl hydrazones, hydrazine, hydrazides moieties, and their underlying mechanisms towards different CRC cell lines, together with a discussion of their limitations and future trends.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2025-01-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143031851","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Chlorogenic Acid Ameliorates CCl4-induced Liver Fibrosis by Modulating the PI3K/AKT/mTOR Autophagy Pathway. 绿原酸通过调节PI3K/AKT/mTOR自噬途径改善ccl4诱导的肝纤维化
IF 2.6 4区 医学
Anti-cancer agents in medicinal chemistry Pub Date : 2025-01-21 DOI: 10.2174/0118715206357043250116063202
Amr Negm, Amira A El-Neanaey, Abada El Sayed Khadr, Maher Abd El Naby Kamel, Abd El-Hamid Abdo Ismail, Ibrahim El Tantawy El Sayed, Wael Sobhy Darwish, Mabrouk Attia Abd Eldaim, Raghda Sobhy Okaz, Mohamed Hamdy Bahr, Nihal Almuraikhi, Nermine Beshara, Tamer M Shawky, Ezat A Mersal
{"title":"Chlorogenic Acid Ameliorates CCl4-induced Liver Fibrosis by Modulating the PI3K/AKT/mTOR Autophagy Pathway.","authors":"Amr Negm, Amira A El-Neanaey, Abada El Sayed Khadr, Maher Abd El Naby Kamel, Abd El-Hamid Abdo Ismail, Ibrahim El Tantawy El Sayed, Wael Sobhy Darwish, Mabrouk Attia Abd Eldaim, Raghda Sobhy Okaz, Mohamed Hamdy Bahr, Nihal Almuraikhi, Nermine Beshara, Tamer M Shawky, Ezat A Mersal","doi":"10.2174/0118715206357043250116063202","DOIUrl":"https://doi.org/10.2174/0118715206357043250116063202","url":null,"abstract":"<p><strong>Background: </strong>Liver fibrosis represents a serious risk to global health by impairing quality of life and elevating the chances of hepatocellular carcinoma, while the intricate role of autophagy can either alleviate or worsen fibrosis depending on its functioning.</p><p><strong>Objective: </strong>Herein, we aimed to investigate the therapeutic effect of chlorogenic acid in CCl4-induced hepatic fibrosis and explore the autophagy pathway as the possible molecular target of chlorogenic acid.</p><p><strong>Methods: </strong>Rats were injected with carbon tetrachloride (1ml/kg) to induce liver fibrosis for 10 weeks. In the current study, the liver fibrosis rats were treated daily with chlorogenic acid (20, 40, and 60 mg/kg) for 30 days. Liver function tests, renal function tests, lipid peroxidation, antioxidant enzyme, anti-inflammatory NF-κB level, and autophagy pathway parameters (PI3K, AKT, mTOR, LC3, and Beclin-1) were assessed.</p><p><strong>Results: </strong>CCl4 elevated serum AST and ALT activity, and hepatic malondialdehyde, PI3K, AKT, and mTOR expressions. It decreased LC3, Beclin-1 expression, and hepatic glutathione level. The results indicated that chlorogenic acid treatment ameliorated the hepatic functions. It declined serum AST and ALT activities, improved hepatic GSH concentration, decreased lipid peroxidation, and downregulated PI3K, AKT, and mTOR protein expressions in hepatic tissue. Moreover, chlorogenic acid increased the hepatic expression of LC3 and Beclin-1. It also significantly decreased NF-kB expression.</p><p><strong>Conclusion: </strong>Chlorogenic acid showed promise in reducing liver damage in rats caused by CCl4 by influencing the autophagy process and adjusting levels of antioxidant and inflammatory markers.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2025-01-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143021748","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Effects of Citrus-derived Diosmetin on Melanoma: Induction of Apoptosis and Autophagy Mediated by PI3K/Akt/mTOR Pathway Inhibition. 柑橘源薯蓣皂苷对黑色素瘤的作用:通过抑制PI3K/Akt/mTOR通路诱导细胞凋亡和自噬。
IF 2.6 4区 医学
Anti-cancer agents in medicinal chemistry Pub Date : 2025-01-21 DOI: 10.2174/0118715206360266250115065234
Jie Li, Mingyuan Xu, Nanhui Wu, Fei Wu, Jiashe Chen, Xiaoxiang Xu, Fei Tan, Yeqiang Liu
{"title":"Effects of Citrus-derived Diosmetin on Melanoma: Induction of Apoptosis and Autophagy Mediated by PI3K/Akt/mTOR Pathway Inhibition.","authors":"Jie Li, Mingyuan Xu, Nanhui Wu, Fei Wu, Jiashe Chen, Xiaoxiang Xu, Fei Tan, Yeqiang Liu","doi":"10.2174/0118715206360266250115065234","DOIUrl":"https://doi.org/10.2174/0118715206360266250115065234","url":null,"abstract":"<p><strong>Background: </strong>Diosmetin (DIOS) is a naturally abundant flavonoid and possesses various biological activities that hold promise as an anti-cancer agent. However, the anti-cancer activities and underlying mechanism of DIOS on cutaneous melanoma remain unclear.</p><p><strong>Objective: </strong>This study seeks to explore the anti-tumor effect and mechanism of DIOS in cutaneous melanoma.</p><p><strong>Methods: </strong>Here, a variety of in vitro and in vivo experiments, combined with RNA sequencing (RNA-seq), were employed to ascertain the potential anti-cutaneous melanoma capacity and mechanism of DIOS.</p><p><strong>Results: </strong>The results demonstrated that DIOS considerably impeded cell proliferation and triggered cell apoptosis in a dose- and time-dependent manner. Concurrently, DIOS markedly elevated the expression of pro-apoptotic proteins (Cleaved caspase-3, Bax, Cleaved PARP, and Cleaved caspase-9) and downregulated the expression of Bcl-2. Additionally, DIOS markedly upregulated the protein expressions of LC3B-II and Atg5, while downregulating p62 protein expression. Notably, pre-treatment with an autophagy inhibitor significantly inhibited DIOSinduced cell apoptosis and autophagy. Mechanistically, DIOS was identified to repress the PI3K/Akt/mTOR signaling pathway by western blot analyses and RNA-seq. Finally, in vivo experiments using a syngeneic mouse model confirmed the anti-tumor effect of DIOS, which exhibited high levels of apoptosis and autophagy.</p><p><strong>Conclusion: </strong>These findings propose that DIOS acts as a potential melanoma therapy that exerts its anti-tumor effects by triggering apoptosis and autophagy via inhibition of the PI3K/Akt/mTOR pathway.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2025-01-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143021816","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Anticancer Properties of Phenylboronic Acid in Androgen-Dependent (LNCaP) and Androgen-Independent (PC3) Prostate Cancer Cells via MAP Kinases by 2D and 3D Culture Methods. 苯基硼酸通过MAP激酶在雄激素依赖性(LNCaP)和雄激素非依赖性(PC3)前列腺癌细胞中的抗癌作用
IF 2.6 4区 医学
Anti-cancer agents in medicinal chemistry Pub Date : 2025-01-20 DOI: 10.2174/0118715206352302241227031015
Duygu Gürsoy Gürgen, Arzu Güneş, Oğuzhan Köse, Arife Ahsen Kaplan, Seda Karabulut, M Başak Tunalı, İlknur Keskin
{"title":"Anticancer Properties of Phenylboronic Acid in Androgen-Dependent (LNCaP) and Androgen-Independent (PC3) Prostate Cancer Cells via MAP Kinases by 2D and 3D Culture Methods.","authors":"Duygu Gürsoy Gürgen, Arzu Güneş, Oğuzhan Köse, Arife Ahsen Kaplan, Seda Karabulut, M Başak Tunalı, İlknur Keskin","doi":"10.2174/0118715206352302241227031015","DOIUrl":"https://doi.org/10.2174/0118715206352302241227031015","url":null,"abstract":"<p><strong>Objective: </strong>This study utilized three cell lines: normal prostate epithelial RWPE-1, androgen-dependent LNCaP, and androgen-independent PC3. We investigated the inhibitory effects of phenylboronic acid (PBA)'s inhibitory effect on cellular proliferation due to its ability to disrupt microtubule formation in prostate cancer cell lines. Additionally, this study aimed to assess the cytotoxic effects of PBA on prostate cancer cells using twodimensional (2D) and three-dimensional (3D) cell culture models.</p><p><strong>Methods: </strong>The IC50 values of PBA and colchicine were determined through viability assays in 2D and 3D models. Colony formation, proliferation, and migration assays were conducted. Immunofluorescence intensity analysis of MAPKKK proteins (ERK, JNK, p38) was performed to explore the mechanism of cellular response to PBA.</p><p><strong>Results: </strong>The IC50 values were determined for each treatment group. After 48-hour of PBA treatment, migration was inhibited more effectively than with colchicine in both cancer cell lines. After 24-hour, PBA reduced colony formation and proliferation. PBA treatment for 24-hour decreased JNK expression in PC3 and LNCaP cells in 2D models. Both PBA and colchicine increased p38 expression in PC3 spheroids. PBA's effects on cell deformation were visualized in semi-thin sections, marking the first ultrastructural observation of PBA-induced morphological defects in cancer cells.</p><p><strong>Conclusion: </strong>PBA exerts antimitotic effects by inhibiting proliferation and migration and triggers diverse metabolic responses across different cell lines. Furthermore the low toxicity of PBA's low toxicity on RWPE-1 cells suggests its potential as a promising chemotherapeutic agent for future studies.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2025-01-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142998834","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The Function of Poly (U) Binding Splicing Factor 60 (PUF60) in Disease Regulation. 聚(U)结合剪接因子60 (PUF60)在疾病调控中的作用
IF 2.6 4区 医学
Anti-cancer agents in medicinal chemistry Pub Date : 2025-01-03 DOI: 10.2174/0118715206346843241119105519
Huijuan Chen, Tian Guan, Jingfeng Song, Yihua Chen
{"title":"The Function of Poly (U) Binding Splicing Factor 60 (PUF60) in Disease Regulation.","authors":"Huijuan Chen, Tian Guan, Jingfeng Song, Yihua Chen","doi":"10.2174/0118715206346843241119105519","DOIUrl":"https://doi.org/10.2174/0118715206346843241119105519","url":null,"abstract":"<p><p>The alternative splicing (AS) of pre-mRNA is an important process in controlling the expression of human genes, which can enrich the diversity of the proteome and regulate gene function. On the contrary, aberrant splicing contributes significantly to numerous human diseases progression, including tumors, neurological diseases, metabolic diseases, infections, and immune diseases. The PUF60, a protein related to RNA splicing, plays critical functions in RNA splicing and gene transcription regulation. In addition, it can achieve synergistic binding with U2AF65 on RNA through interactions in the pyrimidine region, promoting the splicing of introns with weak 3'- splice sites and pyrimidine bundles. Nevertheless, an increasing amount of evidence supports that it shows a significant overexpression pattern in the vast majority of cancer cells and is crucial for embryonic development, indicating that PUF60 may hold the post of a potential therapeutic target for such diseases. These studies have significantly increased our interest in PUF60. Thus, we briefly reviewed the structural domain characteristics of the PUF60, splicing mutants of PUF60, and the roles and functions in human diseases, including various cancers, infections of bacterium and viruses, myositis, and Verheij syndrome. Furthermore, the targeted PUF60 inhibitors and boundedness of the current research were elaborated on in the article. The article effectively communicates critical perception and insight, making it a precious resource for those interested in PUF60 research and treatment.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2025-01-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142942994","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Cucurbitacin E Glucoside as an Apoptosis Inducer in Melanoma Cancer Cells by Modulating AMPK/PGK1/PKM2 Pathway. 葫芦素E糖苷通过调节AMPK/PGK1/PKM2通路在黑色素瘤癌细胞中的凋亡诱导作用
IF 2.6 4区 医学
Anti-cancer agents in medicinal chemistry Pub Date : 2025-01-02 DOI: 10.2174/0118715206345600241216053948
Mohammed Abdalla Hussein, Aya Sayed Sallam, Shaza Ahmed Mohamed, Amera Mahmoud Abdel-Rady, Adam Mostafa Maghrabe, Abdelrahman Wahdan Soltan, Hanan Mohamed Abdelhamid, Gaber E Eldesoky, Seikh Mafiz Alam, Mohammad Shahidul Islam
{"title":"Cucurbitacin E Glucoside as an Apoptosis Inducer in Melanoma Cancer Cells by Modulating AMPK/PGK1/PKM2 Pathway.","authors":"Mohammed Abdalla Hussein, Aya Sayed Sallam, Shaza Ahmed Mohamed, Amera Mahmoud Abdel-Rady, Adam Mostafa Maghrabe, Abdelrahman Wahdan Soltan, Hanan Mohamed Abdelhamid, Gaber E Eldesoky, Seikh Mafiz Alam, Mohammad Shahidul Islam","doi":"10.2174/0118715206345600241216053948","DOIUrl":"https://doi.org/10.2174/0118715206345600241216053948","url":null,"abstract":"&lt;p&gt;&lt;strong&gt;Background: &lt;/strong&gt;Cucurbitacin E glucoside (CEG), a prominent constituent of Cucurbitaceae plants, exhibits notable effects on cancer cell behavior, including inhibition of invasion and migration, achieved through mechanisms such as apoptosis induction, autophagy, cell cycle arrest, and disruption of the actin cytoskeleton.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Objective: &lt;/strong&gt;Melanoma, the fastest-growing malignancy among young individuals in the United States and the predominant cancer among young adults aged 25 to 29, poses a significant health threat. This study aims to elucidate the apoptotic mechanism of CEG against the melanoma cancer cell line (A375).&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Methods: &lt;/strong&gt;The study estimated the IC50 of CEG against the A375 cell line and assessed cell viability, apoptosis, and necrosis upon CEG treatment. Additionally, IC50 values of CEG against Phosphoglycerate kinase1 (PGK1) and Pyruvate Kinase M2 (PKM2) were determined at various levels of concentrations. The impact of CEG on intracellular glutathione (GSH) levels and the activity of key enzymes (GR, SOD, GPx, CAT), as well as markers of apoptosis (P53), and cell cycle regulation (cyclin D1, cyclin E2, cdk2, cdk4), were estimated. Finally, the level of AMP-activated protein kinase (AMPK), PGK1, and PKM2 gene expression levels in A375 cells were also evaluated.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Results: &lt;/strong&gt;The IC50 value of CEG against A375 cells was determined to be 41.87 ± 2.47 µg/mL. A375 cells treated with CEG showed a significant increase in the G0/G1 phase and a decrease in the S and G2/M phases, indicating cell cycle arrest and reduced proliferation. Additionally, there was an increase in the sub-G1 peak, suggesting enhanced apoptosis. Additionally, the pharmacological analysis revealed potent inhibitory activity of CEG against both PGK1 and PKM2 gene expression, with IC50 values 27.89, 11.70, 7.43 and 2.74 µg/mL after incubation periods intervals of 30, 60, 90 and 120 minutes, respectively. In In-Silico study, computational simulations showed a strong binding affinity of CEG towards AMPK, PGK1, and PKM2 activities, with estimated binding energy (∆G) values of -6.5, -7.9, and -8.3 kcal/mol, respectively. Furthermore, incubation of A375 cells with CEG (at concentrations of 20.9, 41.87, and 83.74 µg/mL) led to a significant decrease in GSH levels and the activity of GR, SOD, GPx, CAT, cyclin D1, cyclin E2, cdk2, and cdk4. Notably, CEG treatment upregulated AMPK levels while downregulating PGK1 and PKM2 gene expression significantly.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Conclusion: &lt;/strong&gt;CEG induces apoptosis in melanoma cancer cells (A375) through various mechanisms, including enhanced production of P53 and MDA, inhibition of key enzymes (GR, SOD, GPx, CAT) involved in oxidative stress defense and production of cell cycle regulating enzymes (cyclin D1, cyclin E2, cdk2, cdk4, and upregulation of AMPK and downregulation PGK1, and PKM2 in A375 tumor cells pathways. The downregulation of PKM2 in CEG-treated A375 c","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2025-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142929958","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Novel Celecoxib Derivative, RF26, Blocks Colon Cancer Cell Growth by Inhibiting PDE5, Activating cGMP/PKG Signaling, and Suppressing β-catenin-dependent Transcription. 新型塞来昔布衍生物 RF26 通过抑制 PDE5、激活 cGMP/PKG 信号和抑制β-catenin依赖性转录阻断结肠癌细胞生长
IF 2.6 4区 医学
Anti-cancer agents in medicinal chemistry Pub Date : 2025-01-01 DOI: 10.2174/0118715206318802240821114353
Sara Sigler, Mohammad Abdel-Halim, Reem K Fathalla, Luciana Madeira Da Silva, Adam B Keeton, Yulia Y Maxuitenko, Kristy L Berry, Gang Zhou, Matthias Engel, Ashraf H Abadi, Gary A Piazza
{"title":"Novel Celecoxib Derivative, RF26, Blocks Colon Cancer Cell Growth by Inhibiting PDE5, Activating cGMP/PKG Signaling, and Suppressing β-catenin-dependent Transcription.","authors":"Sara Sigler, Mohammad Abdel-Halim, Reem K Fathalla, Luciana Madeira Da Silva, Adam B Keeton, Yulia Y Maxuitenko, Kristy L Berry, Gang Zhou, Matthias Engel, Ashraf H Abadi, Gary A Piazza","doi":"10.2174/0118715206318802240821114353","DOIUrl":"10.2174/0118715206318802240821114353","url":null,"abstract":"<p><strong>Background: </strong>Previous studies have reported that the cGMP-specific PDE5 isozyme is overexpressed in colon adenomas and adenocarcinomas and essential for colon cancer cell proliferation, while PDE5 selective inhibitors (e.g., sildenafil) have been reported to have cancer chemopreventive activity.</p><p><strong>Aim: </strong>This study aimed to determine the anticancer activity of a novel PDE5 inhibitor, RF26, using colorectal cancer (CRC) cells and the role of PDE5 in CRC tumor growth <i>in vivo</i>.</p><p><strong>Objective: </strong>The objective of this study was to characterize the anticancer activity of a novel celecoxib derivative, RF26, in CRC cells previously reported to lack COX-2 inhibition but have potent PDE5 inhibitory activity.</p><p><strong>Methods: </strong>Anticancer activity of RF26 was studied using human CRC cell lines. Effects on cell growth, cGMPdependent protein kinase (PKG) activity, β-catenin levels, TCF/LEF transcriptional activity, cell cycle distribution, and apoptosis were measured. CRISPR/cas9 PDE5 knockout techniques were used to determine if PDE5 mediates the anticancer activity of RF26 and validate PDE5 as a cancer target.</p><p><strong>Results: </strong>RF26 was appreciably more potent than celecoxib and sildenafil to suppress CRC cell growth and was effective at concentrations that activated PKG signaling. RF26 suppressed β-catenin levels and TCF/LEF transcriptional activity and induced G1 cell cycle arrest and apoptosis within the same concentration range. CRISPR/cas9 PDE5 knockout CRC cells displayed reduced sensitivity to RF26, proliferated slower than parental cells, and failed to establish tumors in mice.</p><p><strong>Conclusion: </strong>Further evaluation of RF26 for the prevention or treatment of cancer and studying the role of PDE5 in tumorigenesis are warranted.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":"52-62"},"PeriodicalIF":2.6,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142118831","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Therapeutic Effects of Crocin Nanoparticles Alone or in Combination with Doxorubicin against Hepatocellular Carcinoma In vitro. Crocin 纳米粒子单独或与多柔比星联合使用对肝细胞癌的体外治疗效果。
IF 2.6 4区 医学
Anti-cancer agents in medicinal chemistry Pub Date : 2025-01-01 DOI: 10.2174/0118715206327654240823074318
Noha S Basuony, Tarek M Mohamed, Doha M Beltagy, Ahmed A Massoud, Mona M Elwan
{"title":"Therapeutic Effects of Crocin Nanoparticles Alone or in Combination with Doxorubicin against Hepatocellular Carcinoma <i>In vitro</i>.","authors":"Noha S Basuony, Tarek M Mohamed, Doha M Beltagy, Ahmed A Massoud, Mona M Elwan","doi":"10.2174/0118715206327654240823074318","DOIUrl":"10.2174/0118715206327654240823074318","url":null,"abstract":"<p><strong>Objective: </strong>Crocin (CRO), the primary antioxidant in saffron, is known for its anticancer properties. However, its effectiveness in topical therapy is limited due to low bioavailability, poor absorption, and low physicochemical stability. This study aimed to prepare crocin nanoparticles (CRO-NPs) to enhance their pharmaceutical efficacy and evaluate the synergistic effects of Cro-NPs with doxorubicin (DOX) chemotherapy on two cell lines: human hepatocellular carcinoma cells (HepG2) and non-cancerous cells (WI38).</p><p><strong>Methods: </strong>CRO-NPs were prepared using the emulsion diffusion technique and characterized by transmission electron microscopy (TEM), scanning electron microscopy (SEM), Zeta potential, and Fourier transform infrared spectroscopy (FT-IR). Cell proliferation inhibition was assessed using the MTT assay for DOX, CRO, CRO-NPs, and DOX+CRO-NPs. Apoptosis and cell cycle were evaluated by flow cytometry, and changes in the expression of apoptotic gene (P53) and autophagic genes (ATG5 & LC3) were analyzed using real-time polymerase chain reaction.</p><p><strong>Results: </strong>TEM and SEM revealed that CRO-NPs exhibited a relatively spherical shape with an average size of 9.3 nm, and zeta potential analysis indicated better stability of CRO-NPs compared to native CRO. Significantly higher antitumor effects of CRO-NPs were observed against HepG2 cells (IC<sub>50</sub> = 1.1 mg/ml and 0.57 mg/ml) compared to native CRO (IC<sub>50</sub> = 6.1 mg/ml and 3.2 mg/ml) after 24 and 48 hours, respectively. Annexin-V assay on HepG2 cells indicated increased apoptotic rates across all treatments, with the highest percentage observed in CRO-NPs, accompanied by cell cycle arrest at the G2/M phase. Furthermore, gene expression analysis showed upregulation of P53, ATG5, and LC3 genes in DOX/CRO-NPs co-treatment compared to individual treatments. In contrast, WI38 cells exhibited greater sensitivity to DOX toxicity but showed no adverse response to CRONPs.</p><p><strong>Conclusion: </strong>Although more in vivo studies in animal models are required to corroborate these results, our findings suggest that CRO-NPs can be a potential new anticancer agent for hepatocellular carcinoma. Moreover, they have a synergistic effect with DOX against HepG2 cells and mitigate the toxicity of DOX on normal WI38 cells.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":"194-206"},"PeriodicalIF":2.6,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142456294","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The Dual Role of ADAMTS9-AS1 in Various Human Cancers: Molecular Pathogenesis and Clinical Implications. ADAMTS9-AS1在多种人类癌症中的双重作用:分子发病机制和临床意义。
IF 2.6 4区 医学
Anti-cancer agents in medicinal chemistry Pub Date : 2025-01-01 DOI: 10.2174/0118715206359325241119075640
Haodong He, Jingjie Yang, Yan Zhou, Xinyan Zheng, Lihan Chen, Zhujun Mao, Chuyuan Liao, Tongtong Li, Haoran Liu, Gang Zhou, Houdong Li, Chengfu Yuan
{"title":"The Dual Role of ADAMTS9-AS1 in Various Human Cancers: Molecular Pathogenesis and Clinical Implications.","authors":"Haodong He, Jingjie Yang, Yan Zhou, Xinyan Zheng, Lihan Chen, Zhujun Mao, Chuyuan Liao, Tongtong Li, Haoran Liu, Gang Zhou, Houdong Li, Chengfu Yuan","doi":"10.2174/0118715206359325241119075640","DOIUrl":"10.2174/0118715206359325241119075640","url":null,"abstract":"<p><p>Long non-coding RNA (lncRNA) is a type of non-coding RNA distinguished by a length exceeding 200 nucleotides. Recent studies indicated that lncRNAs participate in various biological processes, such as chromatin remodeling, transcriptional and post-transcriptional regulation, and the modulation of cell proliferation, death, and differentiation, hence influencing gene expression and cellular function. ADAMTS9-AS1, an antisense long non-coding RNA situated on human chromosome 3p14.1, has garnered significant interest due to its pivotal involvement in the advancement and spread of diverse malignant tumors. ADAMTS9-AS1 functions as a competitive endogenous RNA (ceRNA) that interacts with multiple microRNAs (miRNAs) and plays a crucial role in regulating gene expression and cellular functions by modulating essential signaling pathways, including PI3K/AKT/mTOR, Wnt/β-catenin, and Ras/MAPK pathways. Dysregulation of this factor has been linked to tumor development, migration, invasion, and resistance to apoptotic mechanisms, including as iron-induced apoptosis, underscoring its intricate function in cancer pathology. While current research has clarified certain pathways involved in cancer formation, additional clinical and <i>in vivo</i> investigations are necessary to enhance comprehension of its specific involvement across various cancer types. This review encapsulates the recent discoveries on the correlation of ADAMTS9-AS1 with numerous malignancies, clarifying its molecular mechanisms and its prospective role as a therapeutic target in oncology. Furthermore, it identifies ADAMTS9-AS1 as a potential early diagnostic biomarker and therapeutic target, offering novel opportunities for targeted intervention in oncology.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":"533-543"},"PeriodicalIF":2.6,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142942992","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Emerging Claudin18.2-targeting Therapy for Systemic Treatment of Gastric Cancer: Seeking Nobility Amidst Danger. 用于胃癌全身治疗的新兴 Claudin18.2 靶向疗法:险中求胜。
IF 2.6 4区 医学
Anti-cancer agents in medicinal chemistry Pub Date : 2025-01-01 DOI: 10.2174/0118715206329892240927081033
Xueshuai Ye, Yongqiang Wu, Haiqiang Zhang
{"title":"Emerging Claudin18.2-targeting Therapy for Systemic Treatment of Gastric Cancer: Seeking Nobility Amidst Danger.","authors":"Xueshuai Ye, Yongqiang Wu, Haiqiang Zhang","doi":"10.2174/0118715206329892240927081033","DOIUrl":"10.2174/0118715206329892240927081033","url":null,"abstract":"<p><p>Gastric cancer in advanced stages lacked effective treatment options. claudin18.2 (CLDN18.2) is a membrane protein that is crucial for close junctions in the differentiated epithelial cells of the gastric mucosa, playing a vital role in barrier function, and can be hardly recognized by immune cells due to its polarity pattern. As the polarity of gastric tumor cells changes, claudin18.2 is exposed on the cell surface, resulting in immune system recognition, and making it an ideal target. In this review, we summarized the expression regulation mechanism of claudin18.2 both in normal cells and malignant tumor cells. Besides, we analyzed the available clinical results and potential areas for future research on claudin18.2-positive gastric cancer and claudin18.2-targeting therapy. In conclusion, claudin18.2 is an ideal target for gastric cancer treatment, and the claudin18.2-targeting therapy has changed the treatment pattern of gastric cancer.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":"223-231"},"PeriodicalIF":2.6,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142370794","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
相关产品
×
本文献相关产品
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术官方微信